European Journal of Medicinal Chemistry 78 (2014) 207e216

Contents lists available at ScienceDirect

European Journal of Medicinal Chemistry journal homepage: http://www.elsevier.com/locate/ejmech

Original article

Synthesis, characterization and pharmacological screening of some novel 5-imidazopyrazole incorporated polyhydroquinoline derivatives Piyush N. Kalaria, Shailesh P. Satasia, Dipak K. Raval* Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar 388 120, Gujarat, India

a r t i c l e i n f o

a b s t r a c t

Article history: Received 8 July 2013 Received in revised form 6 September 2013 Accepted 8 February 2014 Available online 19 March 2014

A new category of polyhydroquinoline derivatives 8aet were synthesized in moderate to good yield (64e85%) by one-pot three-component cyclocondensation reaction of 5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1Hpyrazole-4-carbaldehyde 3 with various enaminones 6aeh and different active methylene compounds (malononitrile 7a, ethylcaynoacetate 7b and caynoacetamide 7c) in absolute ethanol. The newly synthesized compounds were evaluated for their in vitro antimalarial activity against Plasmodium falciparum, in vitro antibacterial activity against a panel of pathogenic strains of bacteria and fungi and also for their antitubercular activity against Mycobacterium tuberculosis H37Rv strain. Two of them (8n, 8t) exhibited excellent antimalarial activity. Some of them exhibited excellent antibacterial activity and moderate antituberculosis activity compared with the first line drugs. Ó 2014 Elsevier Masson SAS. All rights reserved.

Keywords: Multicomponent reaction 5-Imidazopyrazole incorporated polyhydroquinolines Antimicrobial activity Antituberculosis activity Antimalarial activity

1. Introduction Malaria is presently prevalent in a broad band around the equator, in areas of the Americas, many parts of Asia, and much of the Africa; however, it is in sub-Saharan Africa where 85e90% of malaria fatalities occur [1]. The World Health Organization has estimated that in 2010, 216 million documented cases of malaria were reported. Around 655,000 people died from the disease, most of them were children in Africa http://en.wikipedia.org/wiki/ Malaria [2]. The emergence and spread of drug-resistant malarial parasites further intensify this serious situation and has compounded the need for the development of new, cost effective antimalarial. Similarly tuberculosis is the second most common reason of death from infectious disease after those due to HIV/AIDS http://en.wikipedia.org/wiki/Tuberculosis [3]. One third of the world’s population is suspected to have been infected with Mycobacterium tuberculosis, with new infections occurring at a rate of about one per second http://en.wikipedia.org/wiki/Tuberculosis [4]. In 2010, there were an estimated 8.8 million new cases and 1.5 million associated deaths mostly found in developing countries http://en.wikipedia.org/wiki/Tuberculosis [5]. In this context, it was thought worth to synthesize various novel compounds which may

* Corresponding author. E-mail addresses: [email protected], [email protected] (D.K. Raval). http://dx.doi.org/10.1016/j.ejmech.2014.02.015 0223-5234/Ó 2014 Elsevier Masson SAS. All rights reserved.

have combined applications as antimalarial, antituberculosis and antimicrobial agents. Among the heterocyclic motifs, azoles constitute enormously important members due to their presence in a multitude of bioactive natural products as privileged pharmacophores. Imidazole and its derivatives are of great significance due to their important roles in biological systems such as antimicrobial [6], anti-inflammatory [7], antitubercular [8], analgesic, anticonvulsant [9], anticancer and anti-parkinson [10] activities. Pyrazole containing compounds have received significant interest owing to their various chemotherapeutic potentials including versatile antineoplastic activities. Many pyrazole derivatives are reported to have the broad spectrum of biological activities, such as antimicrobial [11], anti-inflammatory [12e14], antifungal [15], antitumour, cytotoxic, molecular modelling [16e18] and antiviral [19,20] activities. Much interest has been dedicated to the synthesis of polyhydroquinoline compounds due to their diverse therapeutic and pharmacological properties, such as antitumour, antiatherosclerotic, vasodilator, geroprotective, bronchodilator and hepatoprotective activity [21]. The polyhydroquinoline ring is one of the important heterocyclic core constitutes and prominent structural motif found in numerous pharmaceutically active compounds. Specifically 4-substituteted 1, 4-dihydropyridines (1, 4-DHPs) are well known as Ca2þ channel blockers and have emerged as one of the most important class of drugs for the treatment of

208

P.N. Kalaria et al. / European Journal of Medicinal Chemistry 78 (2014) 207e216

cardiovascular diseases [22]. In addition to the diverse biological activities of polyhydroquinoline, other heterocycles in association with polyhydroquinoline play an essential role in numerous biological processes and possess significant chemical and pharmacological importance. In view of the above biological importance, our aim was to develop new approaches for a variety of heterocycles incorporating polyhydroquinoline scaffold. We attempted to involve pyrazole and imidazole in one molecule which may play a vital role as an important creation part in title compounds. In continuation to our research work [23], we report herein on the utility of 5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazole-4-carbaldehyde as the building block for the synthesis of functionalized polyhydroquinoline scaffold. Study on their pharmacological broadcast in order to get some innovative compounds that could be optimized for potent antimicrobial, antimalarial and antituberculosis agents is also included. 2. Chemistry The synthesis of targeted 5-imidazopyrazole incorporated polyhydroquinoline derivatives are summarized in Scheme 1.

The starting material 5-chloro-3-methyl-1-phenyl-1H-pyrazole-4carbaldehyde 1 was prepared according to VilsmeiereHaack reaction of 3-methyl-1-phenyl-1H-pyrazol-5(4H)-one [24]. The final aldehyde 5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazole-4carbaldehyde 3 was prepared by refluxing compound 1 and imidazole in presence of anhydrous K2CO3 as basic catalyst in DMF as solvent. The required enaminones 6aeh were prepared by the reaction of b-diketones 4aeb with various amines under aqueous condition. The final molecules substituted 4-(5-(1H-imidazol-1-yl)3-methyl-1-phenyl-1H-pyrazol-4-yl)-2-amino-1-phenyl-4,6,7,8tetrahydroquinolin-5(1H)-one were synthesized by refluxing the mixture of 5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazole4-carbaldehyde 3, various enaminones 6aeh and different active methylene compounds (malononitrile 7a, ethylcayno acetate 7b and caynoacetamide 7c) in absolute ethanol using piperidine as the basic catalyst (Scheme 1). The formation of quinoline derivatives 8aet was attempted through an in situ initial formation of heterylidenenitrile, containing the electron-poor C]C double bond, from the Knoevenagel condensation between aldehyde 3 and various active methylene compounds 7aec by loss of water molecules. Finally, Michael addition of enaminones 6aeh to the heterylidene

Scheme 1. Synthesis of the substituted 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4-yl)-2-amino-1-phenyl-4,6,7,8-tetrahydroquinolin-5(1H)-one 8aet. (i) DMF, K2CO3, Reflux 2 h (ii) PEG 600: deionized water (1:1), Reflux 0.5e1 h (iii) Ethanol, Piperidine, Reflux 1e3 h.

P.N. Kalaria et al. / European Journal of Medicinal Chemistry 78 (2014) 207e216

olefins gave acyclic intermediate which underwent cyclization by nucleophilic attack of the NH group on the cyano carbon, followed by tautomerization to afford cyclized polyhydroquinoline derivatives 8aet. 3. Pharmacology 3.1. In vitro antimicrobial activity The minimal inhibitory concentration (MIC) of all synthesized compounds 8aet was determined by broth microdilution method according to National Committee for Clinical Laboratory Standards (NCCLS) [25]. Antibacterial activity was screened against three Gram positive (Streptococcus pneumoniae MTCC 1936, Bacillus subtilis MTCC 441 and Clostridium tetani MTCC 449) and three Gram negative (Escherichia coli MTCC 443, Salmonella typhi MTCC 98, Vibrio cholerae MTCC 3906) bacteria by using ampicillin, ciprofloxacin, norfloxacin and chloramphenicol as the standard antibacterial drugs. Antifungal activity was screened against two fungal species (Candida albicans MTCC 227 and Aspergillus fumigats MTCC 3008) where nystatin and griseofulvin were used as the standard antifungal agents. The strains employed for the activity were procured from the Institute of Microbial Technology, Chandigarh (MTCC-Micro Type Culture Collection). Mueller Hinton broth was used as nutrient medium to grow and dilute the drug suspension for the test. 2% aqueous DMSO was used as the diluent to get the desired concentration of compound to test upon the standard bacterial strains. The results of antimicrobial screening data are shown in Table 1. Table 1 In vitro antimicrobial activity (MIC, mg/mL) of compounds 8aet. Entry

8a 8b 8c 8d 8e 8f 8g 8h 8i 8j 8k 8l 8m 8n 8o 8p 8q 8r 8s 8t A B C D E F

Gram positive bacteria

Gram negative bacteria

Fungi

S.P.

B.S.

C.T.

E.C.

S.T.

V.C.

C.A.

A.F.

MTCC

MTCC

MTCC

MTCC

MTCC

MTCC

MTCC

MTCC

1936

441

449

443

98

3906

227

3008

125 100 250 125 125 200 200 250 250 500 500 200 125 200 200 125 100 100 250 250 100 25 10 50 n.t. n.t.

100 250 250 200 100 200 250 62.5 200 250 250 125 100 500 500 200 125 125 200 200 250 50 100 50 n.t. n.t.

100 250 200 125 125 200 500 200 250 500 200 125 200 200 250 200 100 250 200 200 250 100 50 50 n.t. n.t.

200 125 200 500 100 250 200 200 250 100 250 200 250 200 200 125 200 100 62.5 200 100 25 10 50 n.t. n.t.

62.5 100 200 500 100 250 250 200 125 125 200 250 250 250 200 200 100 200 200 100 100 25 10 50 n.t. n.t.

250 100 250 250 200 500 250 200 250 100 125 250 125 62.5 125 200 250 250 200 500 100 25 10 50 n.t. n.t.

1000 1000 >1000 >1000 >1000 250 500 500 500 1000 250 250 1000 1000 250 500 1000 1000 500 1000 n.t.a n.t. n.t. n.t. 100 500

1000 >1000 250 500 500 1000 1000 >1000 1000 500 500 1000 500 500 1000 500 500 1000 500 1000 n.t. n.t. n.t. n.t. 100 100

S.P.: Streptococcus pneumoniae, B.S.: Bacillus subtilis, C.T.: Clostridium tetani, E.C.: Escherichia coli, S.T.: Salmonella typhi, V.C.: Vibrio cholerae, C.A.: Candida albicans, A.F.: Aspergillus fumigatus, MTCC: Microbial Type Culture Collection. A: Ampicillin, B: Ciprofloxacin, C: Norfloxacin, D: Chloramphenicol, E: Nystatin, F: Griseofulvin. The bold characters indicate the higher or equal activity compared to standard drugs. a n.t.: not tested.

209

3.2. In vitro antituberculosis activity The cheering results from the antibacterial activity impelled us to opt for preliminary screening of the title compounds for their in vitro antituberculosis activity. Primary screening of all the newly synthesized compounds 8aet was conducted at 250 mg/mL and 100 mg/mL against M. tuberculosis H37Rv strain by using LowensteineeJensen medium (conventional method) as described by Rattan [26]. The obtained results are presented in Table 2 in the form of % inhibition. Rifampicin and Isoniazid were used as the standard drugs. 3.3. In vitro antimalarial activity All the synthesized title compounds 8aet were evaluated for their in vitro antimalarial activity against Plasmodium falciparum strain using chloroquine and quinine as the reference compounds. The consequences of the antimalarial screening are expressed as the drug concentration resulting in 50% inhibition (IC50) of parasite growth and are listed in Table 3. 4. Results and discussion 4.1. Analytical results The structures of all the synthesized compounds were confirmed by 1H NMR, FT-IR, 13C NMR, mass and elemental analysis. The 1H NMR spectra of compound 8aet exhibited the presence of the eCH (C4 of polyhydroquinoline ring) proton as a sharp singlet around d 4.32e4.88 ppm and one more singlet arising at d 7.77e7.95 ppm due to imidazole proton (NeCHeN). The aromatic protons resonate as multiplets at around d 6.93e 7.70 ppm. The IR spectrum of compounds 8aet exhibited characteristic absorption band around 1663e1646 cm1 which was attributed to the presence carbonyl group (C5 of polyhydroquinoline ring). The strong absorption band was observed in the range of 1369e1376 cm1 due to eCH3 stretching. The characteristic absorption band in the range 3459e3333 cm1 may be attributed to asymmetric and symmetric stretching of e NH2. In 13C NMR spectra, the carbonyl carbon (C5 of polyhydroquinoline ring) was displayed at d 195.4e196.9 ppm. The signals at around d 57.9e78.5 ppm, 78.3e78.6 ppm and 76.3e 76.8 ppm were assigned to carbon attached to carbonitrile, ester and amide group respectively. The mass spectrum of all the compounds showed molecular ion peak at Mþ corresponding to

Table 2 In vitro antituberculosis activity (% inhibition) of compounds 8aet against M. tuberculosis H37Rv (at concentration 250 mg/mL and 100 mg/mL). Entry

8a 8b 8c 8d 8e 8f 8g 8h 8i 8j 8k

% Inhibition

Entry

250 mg/mL

100 mg/mL

90 12 35 84 63 75 94 20 70 33 45

80 6 30 76 58 66 94 12 61 28 37

8l 8m 8n 8o 8p 8q 8r 8s 8t Rifampicin Isoniazid

% Inhibition 250 mg/mL

100 mg/mL

7 45 38 12 84 91 25 41 67 98 99

4 38 34 9 75 88 20 37 60 98 99

The bold characters indicate the higher or equal activity compared to standard drugs.

210

P.N. Kalaria et al. / European Journal of Medicinal Chemistry 78 (2014) 207e216

Table 3 In vitro antimalarial activity of compounds 8aet. Entry

IC50 (mg/mL)

Entry

IC50 (mg/mL)

8a 8b 8c 8d 8e 8f 8g 8h 8i 8j 8k

1.073 1.79 1.025 0.073 0.057 0.059 1.23 0.096 0.057 1.082 1.47

8l 8m 8n 8o 8p 8q 8r 8s 8t Chloroquine Quinine

0.62 0.71 0.034 0.072 0.57 0.086 1.49 0.067 0.033 0.020 0.268

The bold characters indicate the higher or equal activity compared to standard drugs.

their molecular weights, which confirmed the respective chemical structures. 4.2. Biological section 4.2.1. Antibacterial activity The analysis of antibacterial screening data (Table 1) revealed that all the compounds 8aet showed moderate to very good inhibitory activity. The compound 8h (R2 ¼ OH, X ¼ COOEt) showed maximum activity i.e. 62.5 mg/mL against B. subtilis. Majority of the compounds showed excellent activity against gram positive bacteria B. subtilis and C. tetani as compared to ampicillin (MIC ¼ 250 mg/mL), while compounds 8b (R2 ¼ CH3, X ¼ COOEt), 8c (R2 ¼ CH3, X ¼ CONH2), 8g (R2 ¼ OH, X ¼ CN), 8j (R2 ¼ H, X ¼ CN), 8k (R2 ¼ H, X ¼ COOEt) and 8b (R2 ¼ CH3, X ¼ COOEt), 8i (R2 ¼ OH, X ¼ CONH2), 8q (R2 ¼ OCH3, X ¼ CN), 8r (R2 ¼ OCH3, X ¼ COOEt) showed similar activity as that of the standard drugs against B. subtilis and C. tetani, at 250 mg/mL concentration. The compounds 8b (R2 ¼ CH3, X ¼ COOEt), 8q (R2 ¼ OCH3, X ¼ CN), and 8r (R2 ¼ OCH3, X ¼ COOEt) displayed comparatively good activities i.e. 100 mg/mL against S. pneumonia. The compounds 8a (R2 ¼ CH3, X ¼ CN), 8n (R3 ¼ OH, X ¼ CN) and 8s (R1 ¼ H, R2 ¼ OH, X ¼ COOEt) illustrated highest activity in inhibiting gram negative bacteria i.e. 62.5 mg/mL against S. typhi, V. cholera and E. coli. Compounds 8b (R2 ¼ CH3, X ¼ COOEt), 8e (R2 ¼ Cl, X ¼ COOEt) and 8j (R2 ¼ H, X ¼ CN) also showed same potency against all gram negative bacteria. While compounds 8q (R2 ¼ OCH3, X ¼ CN), 8t (R1 ¼ H, R2 ¼ OH, X ¼ CONH2) and 8r (R2 ¼ OCH3, X ¼ COOEt) showed same potency as that of standard drugs against S. typhi and E. coli. Remaining other compounds are moderate or less active against all gram positive and gram negative bacteria. 4.2.2. Antifungal activity The result of antifungal study (Table 1) of the synthesized polyhydroquinoline derivatives revealed that all the compounds have poor activity against A. fumigates. Where as in comparison with standard fungicidal griseofulvin (MIC ¼ 500 mg/mL), compounds 8f (R2 ¼ Cl, X ¼ CONH2), 8k (R2 ¼ H, X ¼ COOEt), 8l (R2 ¼ Cl, X ¼ CONH2) and 8o (R3 ¼ OH, X ¼ COOEt) contributed excellent antifungal activity i.e. 250 mg/mL against C. albicans. While compounds 8g (R2 ¼ OH, X ¼ CN), 8h (R2 ¼ OH, X ¼ COOEt), 8i (R2 ¼ OH, X ¼ CONH2) and 8s (R1 ¼ H, R2 ¼ OH, X ¼ COOEt) showed same potency i.e. 500 mg/mL against C. albicans. All other compounds showed weak antifungal potency than nystatin and griseofulvin.

and 100 mg/mL against tuberculosis H37Rv strain. The bioassay results obtained for the efficacy of all the synthesized analogues against Mycobacterium tuberculosis H37Rv is summarized in Table 2. The outcome of the result revealed that, compounds 8a (R2 ¼ CH3, X ¼ CN), 8g (R2 ¼ OH, X ¼ CN) and 8q (R2 ¼ OCH3, X ¼ CN) were found to possess excellent activity (i.e. 90%, 94% and 91% at 250 mg/mL) against M. tuberculosis H37Rv. Among the above three compounds, the compound 8g (R2 ¼ OH, X ¼ CN) showed brilliant activity at both the concentration i.e. 94% at 250 mg/mL and 100 mg/ mL. While compounds 8d (R2 ¼ Cl, X ¼ CN) and 8p (R3 ¼ OH, X ¼ CONH2) are moderately active against M. tuberculosis H37Rv. All other compounds showed poor inhibition of M. tuberculosis growth. From the above results, it can be concluded that, compound 8q (R2 ¼ OCH3, X ¼ CN) 8g (R2 ¼ OH, X ¼ CN) may become new class of antitubercular agents in future. 4.2.4. Antimalarial activity All the synthesized compounds 8aet were evaluated for their antimalarial activity against chloroquine and quinine sensitive strain of Plasmodium falciparum. All experiments were performed in duplicate and a mean value of IC50 is mentioned in Table 3. As shown in Table 3, the compounds 8d (R2 ¼ Cl, X ¼ CN), 8e (R2 ¼ Cl, X ¼ COOEt), 8f (R2 ¼ Cl, X ¼ CONH2), 8h (R2 ¼ OH, X ¼ COOEt), 8i (R2 ¼ OH, X ¼ CONH2), 8n (R3 ¼ OH, X ¼ CN), 8o (R3 ¼ OH, X ¼ COOEt), 8q (R2 ¼ OCH3, X ¼ CN), 8s (R2 ¼ OH, X ¼ COOEt) and 8t (R1 ¼ H, R2 ¼ OH, X ¼ CONH2) were found to have IC50 in the range of 0.033e0.096 against P. falciparum strain. These compounds displayed marvellous activity against P. falciparum strain as compared to quinine IC50 0.268. Moreover compounds 8n (R3 ¼ OH, X ¼ CN) and 8t (R1 ¼ H, R2 ¼ OH, X ¼ CONH2) were found to possess moderate activity i.e. IC50 0.033 and 0.034 aligned with chloroquine. Remaining all other compounds were found to be less active against P. falciparum strain against chloroquine and quinine as the standard drugs. 4.2.5. Structureeactivity relationship (SAR) The results of the biological evaluation revealed that the activity was considerably affected by various substituents on the aromatic ring present at first position in polyhydroquinoline derivatives. The hydroxyl group present at various positions in phenyl ring played an important role for the divergence in activity [27]. We observed that the presence of hydroxyl group at ortho position showed significant potency against Bacillus subtilis and Clostridium tetani; while at meta position, it showed maximum activity against V. cholerae and C. tetani as well as increased activity against Plasmodium falciparum. The hydroxyl group present at para position offered higher antifungal activity against C. albicans. Chloro substitution at para position on phenyl ring showed excellent antimalarial activity against P. falciparum. Whereas methoxy group present at para position on phenyl ring showed very good antituberculosis activity against Mycobacterium tuberculosis H37Rv as well as showed best antibacterial activity against all 3 g positive bacteria. Carbonitrile, ester and amide groups present at third position in polyhydroquinoline ring were also found responsible for variation in activity. Particularly the carbonitrile group was found accountable for increasing antituberculosis activity against M. tuberculosis H37Rv whereas ester and amide groups were responsible for increased antifungal activity against C. albicans. The strongly active compounds of the series with reference to the standard drugs are shown in Scheme 2. 5. Conclusion

4.2.3. Antituberculosis activity Antituberculosis screening of all the newly synthesized compounds 8aet was conducted at two concentrations i.e. 250 mg/mL

The objective of the present study was to synthesize and examine the antimicrobial, antimalarial and antituberculosis

P.N. Kalaria et al. / European Journal of Medicinal Chemistry 78 (2014) 207e216

211

Scheme 2. Strong antibacterial, antifungal, antitubercular and antimalarial compounds from the series.

activities of some newly functionalized pyrazole incorporated polyhydroquinoline compounds in search of new structural leads looking promising as potent antimicrobial, antimalarial and antituberculosis agents. This synthetic approach allows the incorporation of three potent bioactive nuclei in a single scaffold through an easy way. Except only two compounds, all the compounds were found to be active against B. subtilis and C. tetani and nearly about half of the compounds showed excellent antifungal activity against Candida albicans. The compounds 8a, 8d, 8g, 8p and 8q exhibited good antituberculosis activities. Majority of the compounds showed excellent activity against strains of P. falciparum as compared to quinine. 6. Experimental section 6.1. Chemistry All reactions were performed with commercially available reagents. They were used without further purification. The solvents used were of analytical grade. All reactions were monitored by thin-layer chromatography (TLC) on aluminum plates coated with silica gel 60 F254, 0.25 mm thickness (Merck). Detection of the components was made by exposure to iodine vapours or UV light. Melting points were taken in melting point apparatus mThermoCal10 (Analab Scientific Pvt. Ltd, India) and are uncorrected. The IR spectra were recorded in KBr on a PerkineElmer Spectrum GX FT-IR Spectrophotometer (PerkineElmer, USA) and only the characteristic peaks are reported in cm1. Mass spectra were recorded on Shimadzu LCMS 2010 spectrometer (Shimadzu, Tokyo, Japan) purchased under PURSE program of DST at Sardar Patel University, Vallabh Vidyanagar. 1H and 13C Nuclear Magnetic Resonance spectra were recorded in DMSO-d6 on a Bruker Avance 400F (MHz) spectrometer (Bruker Scientific Corporation Ltd., Switzerland) using residual solvent signal as an internal standard at 400 MHz and 100 MHz respectively. Chemical shifts are reported in parts per million (ppm). Splitting patterns were designated as follows: s, singlet; d, doublet; dd, doublet of doublet and m, multiplet. The elemental analysis was carried out by using PerkineElmer 2400 series-II elemental analyzer (PerkineElmer, USA) and all compounds are within 0.4% of the theoretical compositions. Yields are not optimized. Ampicillin, griseofulvin, isoniazid and nystatin were purchased from local market.

6.1.1. General procedure for the synthesis of 5-(1H-imidazol-1-yl)3-methyl-1-phenyl-1H-pyrazole-4-carbaldehyde (3) 5-Chloro-3-methyl-1-phenyl-1H-pyrazole-4-carbaldehyde 1 (1.1 g, 5 mmol), appropriate imidazole 2 (0.51 g, 7.5 mmol) and anhydrous potassium carbonate (0.6 g, 10 mmol) in dimethylformamide (5 mL) were charged in a 50 mL round bottom flask with mechanical stirrer and condenser. The reaction mixture was refluxed for 2 h and the progress of the reaction was monitored by TLC. After the completion of reaction (as evidenced by TLC), the reaction mixture was cooled to room temperature and then poured into ice cold water (50 mL) with continuous stirring followed by neutralization with 1 N HCl until pH 7. The separated precipitates of 5-(1H-imidazol-1-yl)-3-methyl-1phenyl-1H-pyrazole-4 carbaldehyde 3 were filtered, thoroughly washed with water, dried, and recrystallized from ethanol. Yield 79%; m.p. 204  C; 1H NMR (400 MHz, DMSO-d6): d 2.59 (s, 3H, CH3), 7.07e7.46 (m, 7H, AreH), 7.94 (s, 1H, imidazole), 9.74 (s, 1H, CHO); ESI-MS (m/z): Calcd. 252.2, found 253.0 (Mþ) 6.1.2. Synthesis of the substituted 3-(phenylamino)cyclohex-2enone (substituted enaminones) (6aeh) Cyclohexane e 1,3 e dione or dimedone (10 mmol), substituted amines (10 mmol) and PEG (polyethylene glycol):deionised water (5 mL:5 mL) were charged in a 50 mL round bottom flask with mechanical stirrer and condenser. The reaction mixture was refluxed for 0.5e1 h. After the completion of reaction (checked by TLC), the substituted enaminones (6 a-h) were filtered and washed with deionised water. The further purification was carried out by leaching in equal volume ratio of water and methanol (10:10 mL) to obtain the pure solid sample. 6.1.3. Synthesis of the substituted 4-(5-(1H-imidazol-1-yl)-3methyl-1-phenyl-1H-pyrazol-4-yl)-2-amino-1-phenyl-4,6,7,8tetrahydroquinolin-5(1H)-one (8aet) A 50 mL round bottom flask, fitted with a reflux condenser, was charged with a mixture of 5-(1H-imidazol-1-yl)-3-methyl-1phenyl-1H-pyrazole-4-carbaldehyde 3 (1 mmol), malononitrile or caynoacetamide or ethylcayno acetate (1 mmol) 7aec, substituted enaminones 6aeh (1 mmol), and catalytic amount of piperidine (2e 3 drops) in ethanol (10 mL). The mixture was heated under reflux for 1e3 h and the progress of the reaction was monitored by TLC. After the completion of reaction, the reaction mixture was cooled to room temperature and stirred magnetically for further 10 min. The solid

212

P.N. Kalaria et al. / European Journal of Medicinal Chemistry 78 (2014) 207e216

mass separated was collected by filtration, washed well with ethanol (10 mL) and crystallized from hot chloroform (10 mL). The physicochemical and spectroscopic characterization data of the synthesized compounds 8aet are given below. 6.1.3.1. 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4y l ) - 2 - a m i n o - 7 , 7 - d i m e t h y l - 5 - o x o - 1 - p - t o l y l - 1, 4 , 5 , 6 , 7 , 8 hexahydroquinoline-3-carbonitrile (8a). Yield 80%; m.p. 278  C; IR (KBr, nmax, cm1): 3459 & 3333 (asym. & sym. stretching of e NH2), 2950 (Ar CeH), 2921 (CeH asym. stretching of CH2 group), 2177 (C^N stretching), 1641(C]O stretching), 1371 (eCH3 stretching); 13 C NMR (100 MHz, DMSO-d6) d: 13.2 (CH3), 22.2 (CH3), 27.3, 28.0 (2C, CH3), 28.5 (C4), 32.4 (C(CH3)2), 41.5, 49.9 (2(CH2)2), 58.3 (Ce CN), 108.1, 121.0, 121.9, 122.5, 122.7, 127.8, 129.67, 129.8, 130.7, 132.4, 134.9, 134.9, 138.1, 147.5, 150.8, 151.3 (16C, AreC), 195.7 (C]O); 1H NMR (400 MHz, DMSO-d6): d 0.93 (s, 3H, CH3), 0.95 (s, 3H, CH3), 1.84 (dd, J ¼ 26.8, 17.2 Hz, 2H, CH2), 2.15 (dd, J ¼ 20.4, 16.4 Hz, 2H, CH2), 2.48 (s, 3H, CH3), 2.50 (s, 3H, CH3), 4.03 (s, 2H, NH2), 4.62 (s, 1H, CH), 7.10e7.40 (m, 11H, AreH), 7.84 (s, 1H, imidazole); ESI-MS (m/z): Calcd. 529.26, found 530.4 (Mþ); Anal. Calcd. (%) for C32H31N7O:72.57; H, 5.90; N, 18.51. Found: C, 72.42; H, 5.79; N, 18.69. 6.1.3.2. Ethyl 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4-yl)-2-amino-7,7-dimethyl-5-oxo-1-p-tolyl-1,4,5,6,7,8hexahydroquinoline-3-carboxylate (8b). Yield 79%; m.p. 247  C; IR (KBr, nmax, cm1): 3418 & 3362 (asym. & sym. stretching of eNH2), 2961 (Ar CeH), 2930 (CeH asym. stretching of CH2 group), 1645 (C]O stretching), 1372 (eCH3 stretching); 13C NMR (100 MHz, DMSO-d6) d: 13.5 (CH3), 15.1 (CH3), 21.3 (CH3), 25.3, 27.8 (2C, CH3), 28.8 (C4), 32.2 (C(CH3)2), 41.7, 50.1 (2(CH2)2), 58.6 (OCH2), 76.4 (Ce COOEt), 110.8, 122.4, 122.5, 122.6, 127.4, 129.3, 129.5, 131.2, 132.4, 133.7, 138.4, 139.8, 148.4, 150.7, 152.8, (15C, AreC), 169.3 (COOEt), 196.0 (C]O); 1H NMR (400 MHz, DMSO-d6): d 0.81 (s, 3H, CH3), d 0.86 (s, 3H, CH3), d 0.95 (t, J ¼ 7.2 Hz, 3H, CH3), 1.70 (d, J ¼ 17.6 Hz, 1H, CH2), 1.94 (d, J ¼ 17.2 Hz, 1H, CH2), 2.14 (s, 2H, CH2), 2.41 (s, 3H, CH3), 2.44 (s, 3H, CH3), 3.44 (m, 2H, CH2), 4.84 (s, 1H, CH), 6.70 (s, 2H, NH2), 6.96e7.40 (m, 11H, AreH), 7.86 (s, 1H, imidazole); ESI-MS (m/z): Cacld. 576.28, found 577.2 (Mþ); Anal. Calcd. (%) for C34H36N6O3: C, 70.81; H, 6.29; N, 14.57. Found: C, 70.70; H, 6.17; N, 14.72. 6.1.3.3. 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4y l ) - 2 - a m i n o - 7 , 7 - d i m e t h y l - 5 - o x o - 1 - p - t o l y l - 1, 4 , 5 , 6 , 7 , 8 hexahydroquinoline-3-carboxamide (8c). Yield 71%; m.p. 230  C; IR (KBr, nmax, cm1): 3409 & 3371 (asym. & sym. stretching of e NH2), 2955 (Ar CeH), 2925 (CeH asym. stretching of CH2 group), 1641 (C]O stretching), 1375 (eCH3 stretching); 13C NMR (100 MHz, DMSO-d6) d: 13.5 (CH3), 21.0 (CH3), 26.6, 27.6 (2C, CH3), 28.9 (C4), 32.1 (C(CH3)2), 41.8, 50.1 (2(CH2)2), 78.3 (CeCONH2), 109.6, 117.9, 119.7, 122.4, 128.5, 129.3, 129.9, 130.0, 132.3, 133.4, 136.5, 138.5, 147.9, 149.8, 151.1 (15C, AreC), 171.1 (CONH2), 195.7 (C]O); 1H NMR (400 MHz, DMSO-d6): d 0.84 (s, 3H, CH3), 0.88 (s, 3H, CH3), 1.73 (d, J ¼ 17.2 Hz, 1H, CH2), 1.96 (d, J ¼ 17.2 Hz, 1H, CH2), 2.16 (s, 2H, CH2), 2.44 (s, 3H, CH3), 2.51 (s, 3H, CH3), 4.69 (s, 1H, CH), 5.74 (s, 2H, NH2), 6.73 (s, 2H, NH2), 7.01e7.64 (m, 11H, AreH), 7.91 (s, 1H, imidazole); ESI-MS (m/z): Calcd. 547.27, found 547.9 (Mþ); Anal. Calcd. (%) for C32H33N7O2:C, 70.18; H, 6.07; N, 17.90. Found: C, 70.24; H, 5.95; N, 17.79. 6.1.3.4. 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4yl)-2-amino-1-(4-chlorophenyl)-7,7-dimethyl-5-oxo-1,4,5,6,7,8hexahydroquinoline-3-carbonitrile (8d). Yield 80%; m.p. 285  C; IR (KBr, nmax, cm1): 3432 & 3356 (asym. & sym. stretching of e NH2), 2953 (Ar CeH), 2929 (CeH asym. stretching of CH2 group), 2187

(C^N stretching), 1644(C]O stretching), 1376 (eCH3 stretching); C NMR (100 MHz, DMSO-d6) d: 13.1 (CH3), 27.3, 27.9 (2C, CH3), 28.5 (C4), 32.3 (C(CH3)2), 41.5, 49.8 (2(CH2)2), 58.1 (CeCN), 108.5, 121.0, 121.9, 122.5, 122.7, 127.8, 129.7, 129.8, 130.7, 132.4, 134.9, 135.1, 138.1, 147.5, 150.8, 151.2 (16C, AreC), 195.7 (C]O); 1H NMR (400 MHz, DMSO-d6): d 0.83 (s, 3H, CH3), 0.87 (s, 3H, CH3), 1.74 (d, J ¼ 17.2 Hz, 1H, CH2), 1.93 (d, J ¼ 17.2 Hz, 1H, CH2), 2.11 (s, 2H, CH2), 2.38 (s, 3H, CH3), 4.33 (s, 1H, CH), 5.48 (s, 2H, NH2), 7.08e7.65 (m, 11H, AreH), 7.82 (s, 1H, imidazole); ESI-MS (m/z): Cacld. 549.2, found 550.1 (Mþ), 551.2 (Mþ2); Anal. Calcd. (%) for C34H36N6O3:C, 67.69; H, 5.13; N, 17.82. Found: C, 67.72; H, 5.01; N, 17.67. 13

6.1.3.5. Ethyl 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4-yl)-2-amino-1-(4-chlorophenyl)-7,7-dimethyl-5-oxo1,4,5,6,7,8-hexahydroquinoline-3-carboxylate (8e). Yield 78%; m.p. 234  C; IR (KBr, nmax, cm1): 3412 & 3369 (asym. & sym. stretching of eNH2), 2955 (Ar CeH), 2925 (CeH asym. stretching of CH2 group), 1639 (C]O stretching), 1371 (eCH3 stretching); 13C NMR (100 MHz, DMSO-d6) d: 13.6 (CH3), 14.9 (CH3), 25.0, 27.5 (2C, CH3), 28.8 (C4), 32.1 (C(CH3)2), 41.6, 49.7 (2(CH2)2), 58.5 (OCH2), 76.5 (Ce COOEt), 110.3, 121.9, 122.4, 122.5, 127.5, 128.9, 128.9, 129.1, 130.3, 138.1, 139.5, 139.9, 148.1, 150.1, 153.1, (15C, AreC), 169.1 (COOEt), 196.1 (C]O); 1H NMR (400 MHz, DMSO-d6): d 0.83 (s, 3H, CH3), d 0.88 (s, 3H, CH3), d 0.95 (t, J ¼ 7.2 Hz, 3H, CH3), 1.72 (d, J ¼ 17.6 Hz, 1H, CH2), 1.97 (d, J ¼ 17.2 Hz, 1H, CH2), 2.17 (s, 2H, CH2), 2.40 (s, 3H, CH3), 2.47 (s, 3H, CH3), 3.42 (m, 2H, CH2), 4.88 (s, 1H, CH), 6.76 (s, 2H, NH2), 6.93e7.43 (m, 11H, AreH), 7.89 (s, 1H, imidazole); ESI-MS (m/z): Calcd. 596.2, found 597.0 (Mþ), 598.2 (Mþ2); Anal. Calcd. (%) for C33H33ClN6O3 : C, 66.38; H, 5.57; N, 14.07. Found: C, 66.30; H, 5.44; N, 14.15. 6.1.3.6. 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4yl)-2-amino-1-(4-chlorophenyl)-7,7-dimethyl-5-oxo-1,4,5,6,7,8hexahydroquinoline-3-carboxamide (8f). Yield 72%; m.p. 245  C; IR (KBr, nmax, cm1): 3414 & 3381 (asym. & sym. stretching of eNH2), 2947 (Ar CeH), 2932 (CeH asym. stretching of CH2 group), 1645 (C]O stretching), 1371 (eCH3 stretching); 13C NMR (100 MHz, DMSO-d6) d: 13.5 (CH3), 26.9, 27.67 (2C, CH3), 28.8 (C4), 31.9 (C(CH3)2), 41.6, 49.7 (2(CH2)2),78.3 (CeCONH2), 109.5, 115.5, 119.9, 122.5, 127.7, 128.3, 129.7, 129.9, 129.9, 132.6, 136.0, 138.3, 148.1, 150.3, 150.9 (15C, AreC), 172.2 (CONH2), 195.6 (C]O); 1H NMR (400 MHz, DMSO-d6): d 0.83 (s, 3H, CH3), 0.86 (s, 3H, CH3), 1.75 (d, J ¼ 17.2 Hz, 1H, CH2), 1.93 (d, J ¼ 17.2 Hz, 1H, CH2), 2.13 (s, 2H, CH2), 2.51 (s, 3H, CH3), 4.70 (s, 1H, CH), 5.76 (s, 2H, NH2), 6.75 (s, 2H, NH2), 7.06e7.64 (m, 11H, AreH), 7.92 (s, 1H, imidazole); ESI-MS (m/z): Calcd. 567.2, found 568.0 (Mþ), 569.3 (Mþ2); Anal. Calcd. (%) for C31H30ClN7O2:C, 65.54; H, 5.32; N, 17.26. Found: C, 65.41; H, 5.24; N, 17.38. 6.1.3.7. 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4yl)-2-amino-1-(4-hydroxyphenyl)-7,7-dimethyl-5-oxo-1,4,5,6,7,8hexahydroquinoline-3-carbonitrile (8g). Yield 84%; m.p. 283  C; IR (KBr, nmax, cm1): 3441 & 3361 asym. & sym. stretching of eNH2), 2945 (Ar CeH), 2926 (CeH asym. stretching of CH2 group), 2171 (C^N stretching), 1646(C]O stretching), 1375 (eCH3 stretching). 13 C NMR (100 MHz, DMSO-d6) d: 13.5 (CH3), 27.2, 28.3 (2C, CH3), 28.5 (C4), 32.4 (C(CH3)2), 41.6, 49.8 (2(CH2)2), 58.5 (CeCN), 108.1, 117.5, 118.7, 121.1, 122.6, 122.8, 127.9, 129.8, 130.0, 130.8, 135.3, 138.3, 147.4, 150.9, 151.6, 155.1 (16C, AreC), 195.6 (C]O); 1H NMR (400 MHz, DMSO-d6): d 0.87 (s, 3H, CH3), 0.90 (s, 3H, CH3), 1.77 (d, J ¼ 17.2 Hz, 1H, CH2), 1.94 (d, J ¼ 17.2 Hz, 1H, CH2), 2.15 (s, 2H, CH2), 2.36 (s, 3H, CH3), 4.30 (s, 1H, CH), 5.50 (s, 2H, NH2), 7.13e7.66 (m, 11H, AreH), 7.77 (s, 1H, OH), 7.95 (s, 1H, imidazole); ESI-MS (m/z): Calcd. 531.2, found 532.2 (Mþ); Anal. Calcd. (%) for C31H29N7O2:C, 70.04; H, 5.50; N, 18.44. Found: C, 70.21; H, 5.41; N, 18.49.

P.N. Kalaria et al. / European Journal of Medicinal Chemistry 78 (2014) 207e216

6.1.3.8. Ethyl 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4-yl)-2-amino-1-(4-hydroxyphenyl)-7,7-dimethyl-5-oxo1,4,5,6,7,8-hexahydroquinoline-3-carboxylate (8h). Yield 79%; m.p. 290  C; IR (KBr, nmax, cm1): 3415 & 3363 (asym. & sym. stretching of eNH2), 2961 (Ar CeH), 2922 (CeH asym. stretching of CH2 group), 1641 (C]O stretching), 1372 (eCH3 stretching); 13C NMR (100 MHz, DMSO-d6) d: 13.7 (CH3), 15.1 (CH3), 25.4, 28.0 (2C, CH3), 28.9 (C4), 32.0 (C(CH3)2), 41.9, 50.2 (2(CH2)2), 58.5 (OCH2), 76.3 (Ce COOEt), 111.1, 117.6, 118.5, 122.1, 122.6, 127.4, 129.3, 130.1, 138.3, 139.3, 139.4, 148.0, 150.1, 152.8, 156.9 (15C, AreC), 169.4 (COOEt), 196.2 (C]O); 1H NMR (400 MHz, DMSO-d6): d 0.82 (s, 3H, CH3), d 0.88 (s, 3H, CH3), d 0.93 (t, J ¼ 7.2 Hz, 3H, CH3), 1.69 (d, J ¼ 17.6 Hz, 1H, CH2), 1.96 (d, J ¼ 17.2 Hz, 1H, CH2), 2.13 (s, 2H, CH2), 2.42 (s, 3H, CH3), 2.47 (s, 3H, CH3), 3.44 (m, 2H, CH2), 4.86 (s, 1H, CH), 6.73 (s, 2H, NH2), 6.95e7.42 (m, 11H, AreH), 7.87 (s, 1H, imidazole), 7.93 (s, 1H, OH); MS: ESI-MS (m/z): Calcd. 578.3, found 578.1 (Mþ); Anal. Calcd. (%) for C33H34N6O4:C, 68.49; H, 5.92; N, 14.52. Found: C, 68.41; H, 5.96; N, 14.52. 6.1.3.9. 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4yl)-2-amino-1-(4-hydroxyphenyl)-7,7-dimethyl-5-oxo-1,4,5,6,7,8hexahydroquinoline-3-carboxamide (8i). Yield 73%; m.p. 248  C; IR (KBr, nmax, cm1): 3412 & 3385 (asym. & sym. stretching of eNH2), 2950 (Ar CeH), 2929 (CeH asym. stretching of CH2 group), 1639 (C]O stretching), 1371 (eCH3 stretching); 13C NMR (100 MHz, DMSO-d6) d: 13.7 (CH3), 26.8, 27.8 (2C, CH3), 28.9 (C4), 32.3 (C(CH3)2), 41.9, 50.2 (2(CH2)2),78.6 (CeCONH2), 110.1, 112.6, 114.8, 120.0, 122.7, 130.1, 130.2, 130.6, 131.9, 136.6, 138.2, 148.3, 151.1, 151.4, 155.4 (15C, AreC), 172.4 (CONH2), 195.6 (C]O); 1H NMR (400 MHz, DMSO-d6): d 0.85 (s, 3H, CH3), 0.89 (s, 3H, CH3), 1.76 (d, J ¼ 17.2 Hz, 1H, CH2), 1.94 (d, J ¼ 17.2 Hz, 1H, CH2), 2.14 (s, 2H, CH2), 2.16 (s, 3H, CH3), 4.65 (s, 1H, CH), 5.75 (s, 2H, NH2), 6.75 (s, 2H, NH2),7.12e7.66 (m, 11H, AreH), 7.89 (s, 1H, imidazole), 7.99 (s, 1H, OH); ESI-MS (m/ z): Calcd. 549.2, found 550 (Mþ); Anal. Calcd. (%) for C31H31N7O3:C, 67.74; H, 5.69; N, 17.84. Found: C, 67.69; H, 5.51; N, 17.91. 6.1.4.0. 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4y l ) - 2 - a m i n o - 7 , 7 - d i m e t h y l - 5 - o x o - 1 - p h e n y l - 1, 4 , 5 , 6 , 7 , 8 hexahydroquinoline-3-carbonitrile (8j). Yield 85%; m.p. 289  C; IR (KBr, nmax, cm1): 3429 & 3378 (asym. & sym. stretching of eNH2), 2958 (Ar CeH), 2923 (CeH asym. stretching of CH2 group), 2184(C^N stretching), 1639 (C]O stretching), 1371 (eCH3 stretching); 13C NMR (100 MHz, DMSO-d6) d: 13.3 (CH3), 27.1, 28.3 (2C, CH3), 28.6 (C4), 32.2 (C(CH3)2), 41.4, 49.8 (2(CH2)2), 58.9 (Ce CN), 108.1, 120.0, 120.7, 121.3, 122.3, 122.4, 127.40, 129.5, 129.7, 130.1, 130.5, 134.8, 138.3, 147.1, 150.7, 151.1 (16C, AreC), 195.6 (C]O); 1H NMR (400 MHz, DMSO-d6): d 0.84 (s, 3H, CH3), 0.89 (s, 3H, CH3), 1.73 (d, J ¼ 17.2 Hz, 1H, CH2), 1.95 (d, J ¼ 17.2 Hz, 1H, CH2), 2.16 (s, 2H, CH2), 2.33 (s, 3H, CH3), 4.32 (s, 1H, CH), 5.46 (s, 2H, NH2), 7.05e7.68 (m, 12H, AreH), 7.84 (s, 1H, imidazole); ESI-MS (m/z): Cacld. 515.2, found 515.3 (Mþ); Anal. Calcd. (%) for C31H29N7O:C, 72.21; H, 5.67; N, 19.02. Found: C, 72.42; H, 5.79; N, 18.81. 6.1.4.1. Ethyl 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4-yl)-2-amino-7,7-dimethyl-5-oxo-1-phenyl-1,4,5,6,7,8hexahydroquinoline-3-carboxylate (8k). Yield 82%; m.p. 275  C; IR (KBr, nmax, cm1): 3421 & 3373 (asym. & sym. stretching of eNH2), 2949 (Ar CeH), 2927 (CeH asym. stretching of CH2 group), 1645 (C]O stretching), 1373 (-CH3 stretching); 13C NMR (100 MHz, DMSO-d6) d: 13.2 (CH3), 15.2 (CH3), 25.3, 27.9 (2C, CH3), 28.9 (C4), 32.4 (C(CH3)2), 41.8, 50.0 (2(CH2)2), 58.7 (OCH2), 76.6 (CeCOOEt), 110.1, 118.9, 122.3, 122.4, 122.8, 127.4, 128.9, 129.1, 129.4, 139.1, 139.2, 139.5, 148.8, 151.0, 152.9, (15C, AreC), 169.3 (COOEt), 195.9 (C]O); 1 H NMR (400 MHz, DMSO-d6): d 0.81 (s, 3H, CH3), d 0.89 (s, 3H, CH3), d 0.94 (t, J ¼ 7.2 Hz, 3H, CH3), 1.71 (d, J ¼ 17.6 Hz, 1H, CH2), 1.92

213

(d, J ¼ 17.2 Hz, 1H, CH2), 2.12 (s, 2H, CH2), 2.39 (s, 3H, CH3), 2.45 (s, 3H, CH3), 3.46 (m, 2H, CH2), 4.82 (s, 1H, CH), 6.72 (s, 2H, NH2), 6.98e 7.42 (m, 11H, AreH), 7.85 (s, 1H, imidazole); ESI-MS (m/z): Calcd. 562.3, found 563.3 (Mþ); Anal. Calcd. (%) for C33H34N6O3:C, 70.44; H, 6.09; N, 14.94. Found: C, 70.21; H, 6.00; N, 15.06. 6.1.4.2. 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4y l ) - 2 - a m i n o - 7 , 7 - d i m e t h y l - 5 - o x o - 1 - p h e n y l - 1, 4 , 5 , 6 , 7 , 8 hexahydroquinoline-3-carboxamide (8l). Yield 81%; m.p. 235  C; IR (KBr, nmax, cm1): 3411 & 3389 (asym. & sym. stretching of eNH2), 2948 (Ar CeH), 2932 (CeH asym. stretching of CH2 group), 1639 (C]O stretching), 1372 (eCH3 stretching); 13C NMR (100 MHz, DMSO-d6) d: 13.6 (CH3), 26.8, 27.7 (2C, CH3), 28.9 (C4), 32.2 (C(CH3)2), 41.8, 50.0 (2(CH2)2),78.4 (CeCONH2), 109.7, 119.9, 121.6, 122.6, 127.7, 129.6, 129.8, 130.1, 130.5, 132.1, 136.5, 138.2, 148.1, 150.1, 150.9 (15C, AreC), 172.4 (CONH2), 195.7 (C]O); 1H NMR (400 MHz, DMSO-d6): d 0.82 (s, 3H, CH3), 0.86 (s, 3H, CH3), 1.71 (d, J ¼ 17.2 Hz, 1H, CH2), 1.95 (d, J ¼ 17.2 Hz, 1H, CH2), 2.15 (s, 2H, CH2), 2.47 (s, 3H, CH3), 4.72 (s, 1H, CH), 5.76 (s, 2H, NH2), 6.78 (s, 2H, NH2), 6.98e7.61 (m, 12H, AreH), 7.93 (s, 1H, imidazole); ESI-MS (m/z): Calcd. 533.3, found 534.0 (Mþ); Anal. Calcd. (%) for C31H31N7O2:C, 69.77; H, 5.86; N, 18.37. Found: C, 70.03; H, 6.00; N, 18.18. 6.1.4.3. 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4yl)-2-amino-1-(2-hydroxyphenyl)-7,7-dimethyl-5-oxo-1,4,5,6,7,8hexahydroquinoline-3-carbonitrile (8m). Yield 84%; m.p. 264  C; IR (KBr, nmax, cm1): 3440 & 3352 (asym. & sym. stretching of eNH2), 2953 (Ar CeH), 2915 (CeH asym. stretching of CH2 group), 2180 (C^N stretching), 1640(C]O stretching), 1370 (eCH3 stretching); 13 C NMR (100 MHz, DMSO-d6) d: 13.1 (CH3), 27.3, 28.1 (2C, CH3), 28.4 (C4), 32.5 (C(CH3)2), 41.5, 49.8 (2(CH2)2), 57.9 (CeCN), 108.4, 113.9, 118.3, 121.1, 121.2, 121.4, 122.3, 126.1, 127.3, 129.6, 129.9, 130.3, 134.8, 138.2, 147.7, 150.7, 151.1, 152.1 (18C, AreC), 196.0 (C]O); 1H NMR (400 MHz, DMSO-d6): d 0.86 (s, 3H, CH3), 0.92 (s, 3H, CH3), 1.78 (d, J ¼ 17.2 Hz, 1H, CH2), 1.94 (d, J ¼ 17.2 Hz, 1H, CH2), 2.18 (s, 2H, CH2), 2.35 (s, 3H, CH3), 4.34 (s, 1H, CH), 5.46 (s, 2H, NH2), 7.12e7.64 (m, 11H, AreH), 7.81 (s, 1H, imidazole), 8.12 (s, 1H, OH); ESI-MS (m/ z): Calcd. 531.2, found, 532.2 (Mþ); Anal. Calcd. (%) for C31H29N7O2:C, 70.04; H, 5.50; N, 18.44. Found: C, 69.89; H, 5.41; N, 18.29. 6.1.4.4. 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4yl)-2-amino-1-(3-hydroxyphenyl)-7, 7-dimethyl-5-oxo-1,4,5,6,7,8hexahydroquinoline-3-carbonitrile (8n). Yield 71%; m.p. 270  C; IR (KBr, nmax, cm1): 3429 & 3364 (asym. & sym. stretching of eNH2), 2961 (Ar CeH), 2925 (CeH asym. stretching of CH2 group), 2169 (C^N stretching), 1645(C]O stretching), 1373 (eCH3 stretching); 13 C NMR (100 MHz, DMSO-d6) d: 13.4 (CH3), 27.5, 28.2 (2C, CH3), 28.6 (C4), 32.2 (C(CH3)2), 41.4, 49.7 (2(CH2)2),58.2 (CeCN), 107.6, 108.3, 108.4, 114.7, 121.2, 122.5, 123.1, 127.8, 129.6, 129.9, 130.2, 130.5, 134.7, 138.2, 147.3, 150.7, 151.1, 160.7 (18C, AreC), 195.4 (C] O); 1H NMR (400 MHz, DMSO-d6): d 0.87 (s, 3H, CH3), 0.90 (s, 3H, CH3), 1.76 (d, J ¼ 17.2 Hz, 1H, CH2), 1.96 (d, J ¼ 17.2 Hz, 1H, CH2), 2.19 (s, 2H, CH2), 2.38 (s, 3H, CH3), 4.36 (s, 1H, CH), 5.46 (s, 2H, NH2), 7.09e7.66 (m, 11H, AreH), 7.84 (s, 1H, imidazole), 8.32 (s, 1H, OH); ESI-MS (m/z): Cacld. 531.2, found 532.1 (Mþ); Anal. Calcd. (%) for C31H29N7O2:C, 70.04; H, 5.50; N, 18.44. Found: C, 69.93; H, 5.36; N, 18.25. 6.1.4.5. Ethyl 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4-yl)-2-amino-1-(3-hydroxyphenyl)-7,7-dimethyl-5-oxo1,4,5,6,7,8-hexahydroquinoline-3-carboxylate (8o). Yield 69%; m.p. 282  C; IR (KBr, nmax, cm1): 3417 & 3383 (asym. & sym. stretching of eNH2), 2963 (Ar CeH), 2932 (CeH asym. stretching of CH2 group), 1636 (C]O stretching), 1375 (eCH3 stretching); 13C NMR

214

P.N. Kalaria et al. / European Journal of Medicinal Chemistry 78 (2014) 207e216

(100 MHz, DMSO-d6) d: 13.8 (CH3), 15.0 (CH3), 25.4, 27.8 (2C, CH3), 28.6 (C4), 32.5 (C(CH3)2), 41.7, 50.1 (2(CH2)2), 58.6 (OCH2), 76.5 (Ce COOEt), 107.1, 108.2, 110.9, 116.2, 122.5, 123.0, 127.4, 127.6, 129.6, 129.7, 138.7, 138.9, 139.4, 148.5, 151.1, 153.01, 160.6 (17C, AreC), 169.3 (COOEt), 196.9 (C]O); 1H NMR (400 MHz, DMSO-d6): d 0.80 (s, 3H, CH3), d 0.87 (s, 3H, CH3), d 0.93 (t, J ¼ 7.2 Hz, 3H, CH3), 1.69 (d, J ¼ 17.6 Hz, 1H, CH2), 1.93 (d, J ¼ 17.2 Hz, 1H, CH2), 2.13 (s, 2H, CH2), 2.40 (s, 3H, CH3), 2.44 (s, 3H, CH3), 3.43 (m, 2H, CH2), 4.86 (s, 1H, CH), 6.69 (s, 2H, NH2), 6.93e7.41 (m, 11H, AreH), 7.86 (s, 1H, imidazole), 8.26 (s, 1H, OH); ESI-MS (m/z): Calcd. 578.3, found 579.0 (Mþ); Anal. Calcd. (%) for C33H34N6O4 : C, 68.49; H, 5.92; N, 14.52. Found: C, 68.66; H, 6.11; N, 14.35. 6.1.4.6. 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4yl)-2-amino-1-(3-hydroxyphenyl)-7,7-dimethyl-5-oxo-1,4,5,6,7,8hexahydroquinoline-3-carboxamide (8p). Yield 64%; m.p. 240  C; IR (KBr, nmax, cm1): 3408 & 3369 (asym. & sym. stretching of eNH2), 2949 (Ar CeH), 2933 (CeH asym. stretching of CH2 group), 1642 (C]O stretching), 1372 (eCH3 stretching); 13C NMR (100 MHz, DMSO-d6) d: 13.6 (CH3), 26.7, 27.8 (2C, CH3), 28.9 (C4), 32.3 (C(CH3)2), 42.0, 49.9 (2(CH2)2),78.5 (CeCONH2), 102.5, 107.1, 109.8, 113.9, 120.1, 122.8, 127.6, 129.9, 130.2, 130.3, 131.5, 136.5, 138.4, 147.7, 150.2, 150.9, 156.9 (17C, AreC), 172.5 (CONH2), 195.6 (C]O); 1H NMR (400 MHz, DMSO-d6): d 0.83 (s, 3H, CH3), 0.87 (s, 3H, CH3), 1.73 (d, J ¼ 17.2 Hz, 1H, CH2), 1.93 (d, J ¼ 17.2 Hz, 1H, CH2), 2.13 (s, 2H, CH2), 2.17 (s, 3H, CH3), 4.70 (s, 1H, CH), 5.77 (s, 2H, NH2), 6.77 (s, 2H, NH2),7.11e7.65 (m, 11H, AreH), 7.91 (s, 1H, imidazole), 8.29 (s, 1H, OH); ESI-MS (m/z): Cacld. 549.2, found 550.2 (Mþ); Anal. Calcd. (%) for C31H31N7O3:C, 67.74; H, 5.69; N, 17.84. Found: C, 67.73; H, 5.81; N, 17.63. 6.1.4.7. 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4yl)-2-amino-1-(4-methoxyphenyl)-7,7-dimethyl-5-oxo-1,4,5,6,7,8hexahydroquinoline-3-carbonitrile (8q). Yield 82%; m.p. 278  C; IR (KBr, nmax, cm1): 3450 & 3360 (asym. & sym. stretching of eNH2), 2949 (Ar CeH), 2931 (CeH asym. stretching of CH2 group), 2175 (C^N stretching), 1641(C]O stretching), 1372 (eCH3 stretching); 13 C NMR (100 MHz, DMSO-d6) d: 13.3 (CH3), 27.2, 28.2 (2C, CH3), 28.7 (C4), 32.3 (C(CH3)2), 41.6, 49.8 (2(CH2)2), 55.1 (OCH3), 58.1 (Ce CN), 107.9, 119.4, 121.2, 121.3, 122.2, 122.6, 127.5, 129.6, 130.3, 130.7, 134.9, 138.6, 147.4, 150.7, 151.2, 156.3 (16C, AreC), 195.6 (C]O); 1H NMR (400 MHz, DMSO-d6): d 0.85 (s, 3H, CH3), 0.91 (s, 3H, CH3), 1.75 (d, J ¼ 17.2 Hz, 1H, CH2), 1.93 (d, J ¼ 17.2 Hz, 1H, CH2), 2.15 (s, 2H, CH2), 2.38 (s, 3H, CH3), 3.58 (s, 3H, CH3), 4.33 (s, 1H, CH), 5.52 (s, 2H, NH2), 7.09e7.70 (m, 11H, AreH), 7.84 (s, 1H, imidazole); ESI-MS (m/ z): Calcd. 545.3, found 546.1 (Mþ); Anal. Calcd. (%) for C32H31N7O2:C, 70.44; H, 5.73; N, 17.97. Found: C, 70.23; H, 5.59; N, 18.07. 6.1.4.8. Ethyl 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4-yl)-2-amino-1-(4-methoxyphenyl)-7,7-dimethyl-5-oxo1,4,5,6,7,8-hexahydroquinoline-3-carboxylate (8r). Yield 79%; m.p. 249  C; IR (KBr, nmax, cm1): 3413 & 3369 (asym. & sym. stretching of eNH2), 2959 (Ar CeH), 2930 (CeH asym. stretching of CH2 group), 1644 (C]O stretching), 1375 (eCH3 stretching); 13C NMR (100 MHz, DMSO-d6) d: 13.6 (CH3), 14.9 (CH3), 25.3, 27.9 (2C, CH3), 28.9 (C4), 32.2 (C(CH3)2), 41.9, 49.9 (2(CH2)2), 55.6 (OCH3) 58.7 (OCH2), 76.8 (CeCOOEt), 110.8, 119.9, 120.1, 122.3, 122.9, 127.6, 129.4, 129.5, 138.5, 138.9, 139.4, 148.4, 150.6, 153.1, 157.9 (15C, AreC), 169.2 (COOEt), 196.8 (C]O); 1H NMR (400 MHz, DMSO-d6): d 0.82 (s, 3H, CH3), d 0.85 (s, 3H, CH3), d 0.94 (t, J ¼ 7.2 Hz, 3H, CH3), 1.72 (d, J ¼ 17.6 Hz, 1H, CH2), 1.96 (d, J ¼ 17.2 Hz, 1H, CH2), 2.14 (s, 2H, CH2), 2.39 (s, 3H, CH3), 2.42 (s, 3H, CH3), 3.45 (m, 2H, CH2), 3.55 (s, 3H, CH3), 4.83 (s, 1H, CH), 6.72 (s, 2H, NH2), 6.97e7.44 (m, 11H, AreH), 7.88 (s, 1H, imidazole); ESI-MS (m/z): Calcd. 592.3, found 593.0

(Mþ); Anal. Calcd. (%) for C34H36N6O4:C, 68.90; H, 6.12; N, 14.18. Found: C, 68.81; H, 5.99; N, 14.32. 6.1.4.9. Ethyl 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4-yl)-2-amino-1-(4-hydroxyphenyl)-5-oxo-1,4,5,6,7,8hexahydroquinoline-3-carboxylate (8s). Yield 78%; m.p. 281  C; IR (KBr, nmax, cm1): 3422 & 3362 (asym. & sym. stretching of eNH2), 2964 (Ar CeH), 2926 (CeH asym. stretching of CH2 group), 1643 (C]O stretching), 1369 (-CH3 stretching); 13C NMR (100 MHz, DMSO-d6) d: 13.6 (CH3), 15.2 (CH3), 21.3 (CH2), 28.9 (C4), 30.1, 38.5 (2(CH2)2), 58.6 (OCH2), 76.3 (CeCOOEt), 110.7, 117.2, 118.9, 122.3, 122.6, 127.8, 129.3, 129.5, 138.7, 138.9, 139.8, 147.9, 150.7, 152.8, 156.4 (15C, AreC), 169.1 (COOEt), 196.0 (C]O); 1H NMR (400 MHz, DMSO-d6): d 0.91 (t, J ¼ 7.2 Hz, 3H, CH3), 1.71 (d, J ¼ 17.6 Hz, 1H, CH2), 1.94 (d, J ¼ 17.2 Hz, 1H, CH2), 2.15 (s, 2H, CH2), 2.42 (s, 3H, CH3), 2.45 (s, 3H, CH3), 3.47 (m, 2H, CH2), 4.85 (s, 1H, CH), 6.71 (s, 2H, NH2), 6.94e7.43 (m, 11H, AreH), 7.90 (s, 1H, imidazole), 7.99 (s, 1H, OH); ESI-MS (m/z): Calcd. 550.2, found 550.9 (Mþ); Anal. Calcd. (%) for C31H30N6O4:C, 67.62; H, 5.49; N, 15.26. Found: C, 67.49; H, 5.62; N, 15.20. 6.1.5. 4-(5-(1H-imidazol-1-yl)-3-methyl-1-phenyl-1H-pyrazol-4yl)-2-amino-1-(4-hydroxyphenyl)-5-oxo-1,4,5,6,7,8hexahydroquinoline-3-carboxamide (8t) Yield 73%; m.p. 240  C; IR (KBr, nmax, cm1): 3415 & 3385 (asym. & sym. stretching of eNH2), 2960 (Ar CeH), 2923 (CeH asym. stretching of CH2 group), 1641 (C]O stretching), 1374 (eCH3 stretching); 13C NMR (100 MHz, DMSO-d6) d: 13.6 (CH3), 21.2 (CH2), 28.9 (C4), 30.3, 38.2 (2(CH2)2), 78.5 (CeCONH2), 109.9, 120.0, 112.7, 122.7, 115.1, 130.0, 130.1, 130.3, 155.4, 132.0, 136.7, 138.4, 148.5, 150.9, 151.3 (15C, AreC), 172.6 (CONH2), 195.9 (C]O); 1H NMR (400 MHz, DMSO-d6): d 1.67 (m, 2H, CH2), 1.92 (s, 3H, CH3), 1.98 (t, J ¼ 7.2 Hz, 2H, CH2), 2.95 (t, J ¼ 17.6 Hz, 2H, CH2), 4.76 (s, 1H, CH), 5.82 (s, 2H, NH2), 6.78 (s, 2H, NH2),7.04e7.62 (m, 11H, AreH), 7.95 (s, 1H, imidazole), 7.96 (s, 1H, OH); ESI-MS (m/z): Calcd. 521.2, found 522.0 (Mþ); Anal. Calcd. (%) for C29H27N7O3:C, 66.78; H, 5.22; N, 18.80. Found: C, 66.94; H, 5.06; N, 18.71. 7. Biological evaluation 7.1. In vitro antimicrobial assay The in vitro antimicrobial activity of all synthesized compounds was carried out by broth microdilution method. 2% DMSO in water was used as the diluent to get the desired concentration of compounds to test upon standard bacterial strains. MuellereHinton broth was used as nutrient medium to grow and dilute the compound suspension for the test bacteria. Sabouraud Dextrose broth was used for fungal nutrition. Inoculum size for test strain was adjusted to 108 CFU mL1 by comparing the turbidity. Serial dilutions were prepared in primary and secondary screening. Each synthesized compounds and the standard drugs were diluted obtaining 2000 mg/mL concentration as a stock solution. The drugs which were found to be active in primary screening (i.e. 500, 250 and 200 mg/mL concentrations) were further screened in their second set of dilution at 100, 50, 25 and 12.5 mg/mL concentration against all microorganisms. 10 micro litre suspensions were further inoculated on appropriate media and growth was noted after 24 and 48 h. The control tube containing no antibiotic was instantaneously subcultured (before inoculation) by spreading a loopful evenly over an area of plate of medium suitable for the growth of the test organism. The tubes were then put for incubation at 37  C overnight. The highest dilution preventing appearance of turbidity after spot subculture was considered as minimal inhibitory concentration (MIC, m/L). All the tubes showing no visible growth

P.N. Kalaria et al. / European Journal of Medicinal Chemistry 78 (2014) 207e216

215

(same as control tube) were subcultured and incubated overnight at 37  C. The amount of growth from the control tube before incubation was compared. In this study Ampicillin, Norfloxacin and Chloramphenicol were used as the standard antibacterial drugs. Nystatin and Griseofulvin were used as standard antifungal drugs. The results are summarized in Table 1.

screening of the compounds reported herein. PNK and SPS wish to acknowledge the University Grants Commission e New Delhi, India for meritorious fellowships awarded to them.

7.2. In vitro antituberculosis assay

Supplementary data related to this article can be found at http:// dx.doi.org/10.1016/j.ejmech.2014.02.015.

The antitubercular activity of all synthesized compound against M. tuberculosis H37Rv was performed by LowensteineeJensen method [26] with minor modification where 250 mg/mL and 100 mg/mL dilution of each compound was added to Lowensteinee Jensen medium and then media was uncontaminated by inspissation method. A culture of M. tuberculosis H37Rv growing on LowensteineeJensen medium was harvested in 0.85% saline in bijou bottle. The stock solutions of the title compounds were prepared in DMSO i.e. 250 mg/mL and 100 mg/mL. These tubes were then incubated at 37  C for 24 h followed by streaking of M. tuberculosis H37Rv (5 _ 104 bacilli per tube). The growth of bacilli was seen after 2 weeks, 3 weeks and finally after 4 week of incubation. The tubes having the compounds were compared with control tubes where medium alone was incubated with M. tuberculosis H37Rv. The concentration at which complete inhibition of colonies occurred was taken as active concentration of the tested compound. The standard strain M. tuberculosis H37Rv was tested with known drug isoniazid and rifampicin for comparison purpose. The results were summarized in Table 2. 7.3. In vitro antimalarial assay All the synthesized compounds were screened for their antimalarial activity against the P. falciparum strain. The P. falciparum strain was acquired from Shree R. B Shah Mahavir Super-speciality hospital, Surat, Gujarat, India, and was used in in vitro tests. The P. falciparum strains were cultivated by a modified method described by Trager and Jensen [28]. Compounds were dissolved in DMSO. The final concentration of DMSO used was not toxic and did not interfere with the assay. The antiparasitic effect of the compounds was measured by growth inhibition percentage as described by Carvalho and Krettli [29]. For experimental purposes, the cultures were synchronized with 5% D-sorbitol when the parasites were in the ring stage [30]. The parasitic suspension, consisting of predominately the ring stage parasites, was adjusted to a 1e2% parasitaemia and 2.5% haematocrit in hypoxanthine-free RPMI-1640 culture medium with 10% human plasma and was exposed to 7 concentrations of each compound for a single cycle of parasite growth for 48 h at 37  C. A positive control with reference to standard antimalarial drugs, chloroquine and quinine, in standard concentrations were used in each experiment. The stock solutions were additionally diluted in whole medium (RPMI 1640 plus 10% human serum) to each of the used concentrations. The concentration that inhibited 50% of parasite growth (IC50 value) was determined by interpolation using Microcal Origin software. The blood smears used were read blind and each duplicate experiment was repeated three times. Acknowledgments The authors are thankful to Head, Department of Chemistry, Sardar Patel University for providing research facilities. We are also thankful to Sophisticated Instrumentation Centre for Applied Research and Training (SICART), Vallabh Vidyanagar for FT-IR analysis at concessional rate and Dhanji P. Rajani, Microcare Laboratory, Surat for antimicrobial, antimalarial and antituberculosis

Appendix A. Supplementary data

References [1] The malERA Consultative Group on Drugs, A research agenda for malaria eradication: drugs, PLoS Medicine 8 (1) (2011) e1000402, http://dx.doi.org/ 10.1371/journal.pmed.1000402. [2] WHO, World Malaria Report, 2011. http://www.who.int/malaria/world_ malaria_report_2011/en/. [3] Dolin, G.L. Mandell, J.E. Bennett, Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases, seventh ed., Churchill Livingstone/Elsevier, Philadelphia, PA, 2010, ISBN 978-0-443-06839-3 (Chapter 250). [4] WHO, Tuberculosis Fact Sheet, 2011. [5] WHO, The Sixteenth Global Report on Tuberculosis, 2011. [6] I. Takeuchi, M. Sugiura, K. Yamamoto, T. Ito, Y. Hamada, Yakuga Zasshi 105 (1985) 554e561. [7] F. Suzuki, T. Kuroda, T. Tamura, S. Sato, K. Ohmori, S. Ichikawa, Journal of Medicinal Chemistry 35 (1992) 2863e2870. [8] J. Pandey, V.K. Tiwari, S.S. Verma, V. Chaturvedi, S. Bhatnagar, S. Sinha, A.N. Gaikwad, R.P. Tripathi, European Journal of Medicinal Chemistry 44 (2009) 3350e3355. [9] M. Pinza, Z. Farina, A. Cerri, U. Pfeiffer, M.T. Riccaboni, S. Banfi, R. Biagetti, O. Pozzi, M. Magnani, L. Dorigotti, Journal of Medicinal Chemistry 36 (1993) 4214e4220. [10] H. Miyachi, H. Kiyota, M. Segawa, Bioorganic & Medicinal Chemistry Letters 8 (1998) 1807e1812. [11] A.M. Isloor, B. Kalluraya, P. Shetty, European Journal of Medicinal Chemistry 44 (2009) 3784e3787. [12] A.A. Bekhit, H.M.A. Ashour, Y.S.A. Ghany, A.E.A. Bekhit, A. Baraka, European Journal of Medicinal Chemistry 43 (2008) 456e463. [13] G. Menozzi, L. Masti, P. Fassa, F. Mattioli, M. Ghia, Journal of Heterocyclic Chemistry 34 (1997) 963e968. [14] A.K. Tewari, A. Mishra, Bioorganic and Medicinal Chemistry 9 (2001) 715e 718. [15] O. Prakash, R. Kumar, V. Parkash, European Journal of Medicinal Chemistry 43 (2008) 435e440. [16] M.A.F. Vera-DiVaio, A.C.C. Freitas, H.C.A. Castro, S. de Albuquerque, L.M. Cabral, C.R. Rodrigues, M.G. Albuquerque, R.C.A. Martins, M.G. Henriques, L.R.S. Dias, Bioorganic and Medicinal Chemistry 17 (2009) 295e302. [17] H.E. Skipper, R.K. Robins, J.R. Thompson, Proceedings of the Society for Experimental Biology and Medicine 89 (1955) 589e594. [18] T.C. Hsu, R.K. Robins, C.C. Cheng, Science 13 (1956) 848e868. [19] R. Storer, C.J. Ashton, A.D. Baxter, M.M. Hann, C.L.P. Marr, A.M. Mason, C.L. Mo, P.L. Myers, S.A. Noble, C.R. Penn, N.G. Weir, J.M. Woods, P.L. Coe, Nucleosides, Nucleotides and Nucleic Acids 18 (1999) 203e216. [20] M.J. Genin, C. Biles, B.J. Keiser, S.M. Poppe, S.M. Swaney, W.G. Tarpley, Y. Yagi, D.L. Romero, Journal of Medicinal Chemistry 43 (2000) 1034e1040. [21] A. Sausins, G. Duburs, Heterocycles 7 (1988) 269e289. [22] B. Loev, K.M. Snader, Journal of Organic Chemistry 30 (1965) 1914e1916. [23] (a) J.R. Avalani, D.S. Patel, D.K. Raval, Journal of Chemical Sciences 124 (2012) 1091e1096; (b) B. Thummar, U.P. Tarpada, D.K. Raval, Journal of Heterocyclic Chemistry (2013), http://dx.doi.org/10.1002/jhet.1870; (c) S.P. Satasia, P.N. Kalaria, D.K. Raval, RSC Advances 3 (2013) 3184e3188; (d) J.R. Avalani, D.S. Patel, D.K. Raval, Journal of Molecular Catalysis B: Enzymatic 90 (2013) 70e75; (e) A. Dadhania, V. Patel, D. Raval, Journal of Chemical Sciences 124 (2012) 921e926; (f) D.S. Patel, J.R. Avalani, D.K. Raval, Journal of the Brazilian Chemical Society 23 (2012) 1951e1954; (g) A. Dadhania, V. Patel, D. Raval, Comptes Rendus Chimie 15 (2012) 378e 383; (h) A. Dadhania, V. Patel, D. Raval, Journal of the Brazilian Chemical Society 22 (2011) 511e516; (i) U.P. Tarpada, B.B. Thummar, D.K. Raval, Journal of Saudi Chemical Society. doi: 10.1016/j.jscs.2012.07.014. [24] H.Q. Xiao, G.P. Ouyang, X.D. Sun, X.D. Yao, G.Q. Bao, C.Z. Qi, Synthesis of pyrazoleoxime esters, Chinese Journal of Synthetic Chemistry 13 (2005) 600e602. [25] NCCLS (National Committee for Clinical Laboratory Standards), Performance Standards for Antimicrobial Susceptibility Testing: Twelfth Informational Supplement, 2002, 1-56238-454-6 M100-S12, M7. [26] A. Rattan, in: B.I. Churchill (Ed.), Antimicrobials in Laboratory Medicine, Livingstone, New Delhi, 2000, pp. 85e108.

216

P.N. Kalaria et al. / European Journal of Medicinal Chemistry 78 (2014) 207e216

[27] P. Thapa, R. Karki, M. Yun, T. Kadayat, E. Lee, H. Kwon, Y. Na, W. Cho, N. Kim, B. Jeong, Y. Kwon, E. Lee, European Journal of Medicinal Chemistry 52 (2012) 123e136. [28] W. Trager, J.B. Jensen, Science 193 (1976) 673e675.

[29] L.H. Carvalho, A.U. Krettli, Memórias do Instituto Oswaldo Cruz. 86 (Suppl. II) 181e184. [30] C. Lambros, J.P. van der Berg, Journal of Parasitology 65 (1979) 418e420.

Synthesis, characterization and pharmacological screening of some novel 5-imidazopyrazole incorporated polyhydroquinoline derivatives.

A new category of polyhydroquinoline derivatives 8a-t were synthesized in moderate to good yield (64-85%) by one-pot three-component cyclocondensation...
957KB Sizes 0 Downloads 3 Views